WO2015101794A1 - Pharmaceutical composition - Google Patents

Pharmaceutical composition Download PDF

Info

Publication number
WO2015101794A1
WO2015101794A1 PCT/GB2015/050006 GB2015050006W WO2015101794A1 WO 2015101794 A1 WO2015101794 A1 WO 2015101794A1 GB 2015050006 W GB2015050006 W GB 2015050006W WO 2015101794 A1 WO2015101794 A1 WO 2015101794A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical composition
composition according
nitisinone
saccharide
fat
Prior art date
Application number
PCT/GB2015/050006
Other languages
English (en)
French (fr)
Inventor
James Harrison
Stephen Fuller
Tobias Josef BROWN
Original Assignee
Cycle Pharmaceuticals Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to LTEP15700003.5T priority Critical patent/LT3089740T/lt
Priority to PL15700003T priority patent/PL3089740T3/pl
Priority to DK15700003.5T priority patent/DK3089740T3/da
Priority to EP15700003.5A priority patent/EP3089740B1/en
Priority to US15/109,220 priority patent/US10328029B2/en
Priority to CA2935013A priority patent/CA2935013C/en
Application filed by Cycle Pharmaceuticals Ltd filed Critical Cycle Pharmaceuticals Ltd
Priority to ES15700003T priority patent/ES2806739T3/es
Priority to RS20200849A priority patent/RS60551B1/sr
Priority to SI201531285T priority patent/SI3089740T1/sl
Publication of WO2015101794A1 publication Critical patent/WO2015101794A1/en
Priority to CY20201100659T priority patent/CY1123394T1/el
Priority to HRP20201135TT priority patent/HRP20201135T1/hr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism

Definitions

  • the present invention relates to a pharmaceutical composition.
  • it concerns a pharmaceutical composition of nitisinone which possesses excellent storage stability.
  • Nitisinone (lUPAC name: 2-[2-nitro-4-(trifluoromethyl)benzoyl] cyclohexane- 1 ,3-dione ) is currently approved under the trade name ORFADIN (FDA NDA N021232) to treat Hereditary Tyrosinemia type- 1 (HT-1). This is a rare genetic disorder in which the newborn child is unable to break down the amino acid tyrosine, which causes the build up of toxic metabolites that can lead to liver failure, kidney dysfunction and neurological problems. Daily treatment with nitisinone (dosage 1 mg / kg) prevents the build up of toxic metabolites. itisinone has the structural formul
  • nitisinone is currently administered orally in capsule form, available in three dosages, 2, 5 and 10 mg. It is an immediate release capsule formulation of nitisinone with pre-gelatinised starch as the only excipient.
  • I lowevcr one of the major disadvantages concerning the currently available commercial product is that, due to stability issues, it must be stored at 2-8°C (i.e. it must be refrigerated). This is a significant drawback to HT-1 patients and their families.
  • the benefit of such a formulation is that it will allow 11 1 - 1 patients to travel more freely and to gain more independence without the worry of keeping the medication within the required storage conditions.
  • a pharmaceutical composition suitable for oral administration wherein the composition is in the form of a compressed tablet and comprises nitisinone, or a pharmaceutically acceptable salt thereof, at least one saccharide, wherein the saccharide is a di saccharide or oligosaccharide, and at least one pharmaceutically acceptable excipient, provided that the composition does not contain magnesium stearate.
  • nitisinone is prone to forming cyclisation products at increased temperature.
  • the cyclisation reaction which is shown below, involves cyclisation of one of the carbonyl groups of the cyclohexanedione ring onto the phenyl ring, and loss o the nitro group.
  • the cyclised impurity identified must not exceed the qualification threshold (i.e. below which toxicity testing is not required) of 0.15% w/w (with respect to the total amount of nitisinone) or 1.0 mg/day (whichever is lower) in accordance with ICH ( International Conference on I larmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use) guidelines for drug substance impurities Q3A(R2), when limited to a maximum daily API dose of 2 g/day.
  • the impurity should not exceed a qualification threshold of 0.2% or 0.5% w/w (with respect to the total amount of nitisinone in the composition), depending on the dose of nit isinone to be administered.
  • Possible alternative nitisinone capsule and tablet compositions include those disclosed in EP0591275. These compositions typically contain a number of components, in particular the lubricant magnesium stearate. However, the inventors have found that the presence of magnesium stearate, and metal ions in general, renders the compositions unstable on storage under a range of temperature conditions. Surprisingly, tablet compositions which do not contain magnesium stearate are no longer prone to formation of the cyclised impurity associated with nitisinone, particularly during storage.
  • the composition does not contain metal ions.
  • the composition does not contain pre- gelatinised starch, and preferably it does not contain any starch.
  • the composition may also contain at least one saccharide and/or at least one fat. These additional components may further improve the stability of the present composition.
  • a pharmaceutical composition suitable for oral administration wherein the composition comprises nitisinone, or a pharmaceutically acceptable salt thereof, at least one saccharide, and at least one fat.
  • composition of this aspect may be presented in any physical form. However, it is preferably provided as a compressed dosage form, such as a tablet.
  • a compressed dosage form such as a tablet.
  • the composition of the invention has been shown to exhibit excellent stability under both accelerated and long-term conditions. In particular, very little or no cyclisation product is observed.
  • the tablet form is also beneficial from the patient's perspective, making it easier to take the required medication.
  • a pharmaceutical composition suitable for oral administration comprising nitisinone, or a pharmaceutically acceptable salt thereof, at least one saccharide, wherein the saccharide is a disaccharide or oligosaccharide, and at least one pharmaceutically acceptable excipient, provided that the composition does not contain magnesium stearate or starch.
  • the composition does not contain metal ions.
  • compositions containing magnesium stearate and/or starch have been shown to promote the formation of unwanted impurities (i.e. cyclised product) which breach regulatory guidelines for medicinal products.
  • salts with acids refers to salts of nitisinone and includes salts with organic bases, salts with inorganic acids, salts with organic acids and salts with basic or acidic amino acids. Salts with acids may, in particular, be employed in some instances.
  • Exemplary salts include hydrochloride salt, acetate salt, trifluoroaeetate salt, methanesulfonate salt, 2-hydroxypropane-l ,2,3-tricarboxylate salt, (2R,3R)-2,3- dihydroxysuccinate salt, phosphate salt, sulphate salt, benzoate salt, 2-hydroxy-benzoate salt, S-(+)-mandelate salt, S-(-)-malate salt, S-(-) pyroglutamate salt, pyruvate salt, p- toluenesulfonate salt, l-R-(-)-camphorsulfonate salt, lumarate salt and oxalate salt.
  • the nilisinonc used in the composition of the invention may be in either solvate (e.g. hydrate) or non-solvate (e.g. non-hydrate) form.
  • the saccharide may be a monosaccharide, disaccharide or oligosaccharide.
  • one or more monosaccharides may also be present, in addition to the disaccharide or oligosaccharide.
  • Suitable monosaccharides include glucose (dextrose), fructose (levulose), galactose, ribose, arabinose, allose, fucose, altrose, mannitol, mannose, sorbitol and xylose.
  • Suitable disaccharides include lactose, lactulose, sucrose, sucralose, trehalose, and robinose.
  • Suitable oligosaccharides include saccharides comprising from 3 to 9 monosaccharides, preferably from 3 to 6 monosaccharides, or from 2 to 4 disaccharides, or appropriate combinations thereof.
  • the saccharide is a monosaccharide or disaccharide.
  • the saccharide is a disaccharide, such as lactose (e.g. SuperTab® 30GR).
  • the fat may be a monoglyceride, diglyceride or triglyceride, the fatty acid moieties of which are saturated or unsaturated, preferably saturated, and contain from 6 to 30 carbon atoms. More preferably, the fat is a diglyceride.
  • Such fats provide beneficial properties as lubricants in the preparation of the composition of the invention (particularly where the composition is a tablet). Surprisingly, and in contrast to the lubricant magnesium stearate, these fats have been found to aid stability of the composition by preventing the formation of impurities in a similar manner to the saccharide component.
  • the fatty acid moieties of the fat contain from 18 to 24 carbon atoms, more preferably from 20 to 22 carbon atoms.
  • the fat of the composition is most preferably glycerol dibehenate (e.g. Compritol® 888). This material may provide even further enhanced stability to the composition, specifically in preventing the formation of the aforementioned cyclised product.
  • the term 'saturated' as used herein refers to fatty acid moieties containing only carbon- carbon single bonds, i.e. an alkyl group.
  • the term 'unsaturated' as used herein refers to fatty acid moieties containing at least one carbon-carbon double or triple bond (i.e. an alkenyl group, -Cl fr I h-. or an alkynyl group, -CH ⁇ CH-). Any alkenyl groups which may be present may exist in either cis or trans geometries.
  • the fatty acid moieties of the fat are either saturated, or unsaturated with one or more alkenyl groups.
  • the saccharide is lactose and the fat is glycerol dibehenate.
  • This combination of excipients has been shown to provide a composition which has evident stability in terms of long-term storage (e.g. no degradation of the drug substance), particularly in formulations comprising a compressed tablet.
  • the stability of the present pharmaceutical compositions is an important advantage of the invention. Accordingly, the pharmaceutical compositions of the invention may be characterised such that they have a stability (i.e. up to 0.5% nitisinone impurities, preferably up to 0.2% nitisinone impurities, in particular the cyclised impurity, as measured by HPLC under ICH guidelines) of at least 2 months at room temperature (e.g. 15 to 25 °C), preferably at least 4 months or 6 months, most preferably 12 months at room temperature. Alternatively, the compositions may have a stability of at least 2 months at 25 to 50 °C, preferably at least 4 months or 6 months, most preferably 12 months.
  • a stability i.e. up to 0.5% nitisinone impurities, preferably up to 0.2% nitisinone impurities, in particular the cyclised impurity, as measured by HPLC under ICH guidelines
  • the compositions may have a stability of at least 2 months at 25 to 50 °C,
  • the compositions may exhibit a stability of at least 2 months under accelerated storage conditions of 40 °C and 75% relative humidity, preferably at least 4 months, and/or at least 2 months, preferably at least 4 months, under long-term storage conditions of 25 °C and 60% relative humidity.
  • the compositions may exhibit a stability of at least 2 months under intermediate conditions of 30 °C and 65% relative humidity, preferably at least 4 months or 6 months, most preferably 12 months. More preferably, the composition has a stability of at least 6 months, preferably at least 12 months, under accelerated conditions of 40 °C and 75% relative humidity, and/or at least 6 months, preferably at least 12 months, under long-term conditions of 25 °C and 60% relative humidity. Even more preferably, the composition has a stability such that substantially no increase in cyclised impurity is detectable by HPLC over a period of at least 2 months, preferably at least 4 or 6 months, even more preferably at least 12 months.
  • the level of cyclised product may be determined by means of the HPLC method defined in the examples.
  • the invention provides the pharmaceutical composition according the invention, for use in therapy.
  • the composition of the invention for use in the treatment of tyrosinemia, such as Hereditary Tyrosinemia type- 1 (HT-1), or alkaptonuria.
  • tyrosinemia such as Hereditary Tyrosinemia type- 1 (HT-1)
  • HT-1 Hereditary Tyrosinemia type-1
  • HT-1 Hereditary Tyrosinemia type-1
  • a process for producing nitisinone wherein the process comprises extracting the nitisinone product under basic conditions, such as at a pi I of 8 or more, preferably 9 or more, 10 or more, or 1 1 or more, more preferably 1 1 to 13, e.g. around 12.
  • a process for producing a crystalline form of nitisinone comprising recrystallising a sample of nitisinone from an acetone:water solvent system.
  • the recrystallisation is carried out at a temperature of up to 50 °C, more preferably up to 45 °C.
  • the recrystallisation is carried out using a ratio of acetone: water from 3 :7 to 7:3, more preferably from 3 :4 to 6:3.
  • Figure la shows drug stability data under accelerated conditions for ORFADIN and a tablet formulation according to the invention against a qualification threshold of 0.5%.
  • Figure lb shows drug stability data under accelerated conditions for ORFADIN against a qualification threshold of 0.2%.
  • Figure lc shows dmg stability data under accelerated conditions for a 160 mg lactose capsule formulation as described herein against a qualification threshold of 0.2%.
  • Figure Id shows drug stability data under accelerated conditions for a 160 mg tablet formulation according to the invention against a qualification threshold of 0.2%.
  • Figure le shows drug stability data under accelerated conditions for a 120 mg tablet formulation according to the invention against a qualification threshold of 0.2%.
  • Figure 2a shows drug stability data under long-term conditions for ORFADIN and a tablet formulation according to the invention against a qualification threshold of 0.5%.
  • Figure 2b shows drug stability data under long-term conditions for ORFADIN against a qualification threshold of 0.2%.
  • Figure 2c shows drug stability data under long-term conditions for a 160 mg lactose capsule formulation as described herein against a qualification threshold of 0.2%.
  • Figure 2d shows drug stability data under long-term conditions for a 160 mg tablet formulation according to the invention against a qualification threshold of 0.2%.
  • nitisinone is outlined in the scheme below.
  • adaptations to stage 1 especially the extraction and precipitation steps, and the timing of the treatment of the O-acyl intermediate with TMSCN, improved the yield from less than 50 % to more than 85 %, and maintained a purity of greater than 95 % (compared to the route described in US patent 5,550, 165).
  • Changing the solvent used for the recrystallisation of the product from ethyl acetate, as also described in US patent 5,550,165, to acetone/water improved yields from 37 % to greater than 90 %, whilst maintaining a purity of greater than 99.9 %.
  • the added advantage of using such solvents for the recrystallisation is that they conform to ICH guidelines for residual solvent levels.
  • BA benzoic acid
  • the final recrystallisation includes a filtration through a polishing filter to remove particulates, and so the GMP process would involve filtration of the hot acetone solution.
  • the stability of stage 1 at 45°C was tested over a 6 hour period to confirm that the Nitisinone would not degrade under these conditions (Table 4). No loss of purity was observed. Therefore, acetone could be used in the hot filtration step of the GMP process.
  • BA benzoic acid
  • the final recrystallisation is also excellent for purging remaining substituted benzoic acid to very low levels.
  • the filter-cake was washed with water to remove the residual mother liquors. Due to its high solubility in acetone and other organic solvents, a solvent wash was not appropriate as high yield losses would likely be incurred. A water wash also helped remove residual inorganics. Drying studies were therefore carried out to determine the stability of the recrystallised material and try to ensure that there would be no unexpected degradation during the drying phase of the scale-up manufacture (Table 5). Table 5: Stage 2 drying studies
  • a sample of approximately 150 g of substance was used for the testing programme, with individual samples being approximately 7 g in size.
  • the test was designed to simulate the product package, i.e. the sample was packed in a polythene bag, sealed and then placed inside another polythene bag liner. The sample was then held in a fibreboard cask with a fibreboard lid.
  • the sample was stored in an incubator maintained at 25°C ⁇ 2°C / 60% ⁇ 5 % relative humidity (RH), and 40°C ⁇ 2°C / 75% ⁇ 5 % RH.
  • a number of fillers also accelerated the formation of the cyclised impurity, notably and most significantly pre-gelatinised starch, which is currently used in the approved ORFADIN capsule formulation.
  • the use of lactose did not promote the formation of the cyclised impurity.
  • the excipients and active pharmaceutical ingredient (API) were passed through a 400 ⁇ screen before blending.
  • the tablets were then compressed to a hardness of around 5.5KP (5- 6) using standard curvature 7 mm round tooling.
  • the tablets had a final weight of 120 mg.
  • Glycerol dibehenate in the form of Compritol 888, is a registered GRAS substance with no upper acceptable daily intake (ADI) limit.
  • the amount of glycerol dibehenate used in the formulation is within the amounts previously used in other formulations listed in the FDA inactive ingredients guide (up to 60mg/dose).
  • Tablets (or capsules) of the composition may be contained in high density polyethylene (HDPE) bottles. No desiccant is required in the bottles containing the composition of the invention, since an excipient compatibility study showed that higher water content increased the stability of the product.
  • HDPE high density polyethylene
  • the formulation was developed to stay within specifications under accelerated storage conditions for a period of at least 6 months, which then allowed extrapolation of data from stability testing at the long-term conditions to demonstrate room temperature storage of the drug formulation product. At all stages, the new formulation was directly compared to the commercially available drug product.
  • ORFADIN Example 5 - Capsule vs Tablet Formulation vs ORFADIN
  • a capsule formulation was developed for comparison to the tablet formulation of the same excipient blend, as shown in the table below (Table 10), in order to determine the influence of dose form on stability.
  • excipients and API were passed through a 1000 ⁇ screen before blending in a turbular blender (30 minutes at 30 rpm). Following blending, 160 mg of the blend is placed into size 3 HPMC capsules using an automated encapsulation machine (Bonapace In-cap).
  • the lactose capsule formulation showed significant improvements in stability when compared to ORFADIN; however, projections demonstrated that the amount of cyclised impurity would breach the qualification threshold of 0.5% within 6 months for the 2 mg capsules. For the 0.2% qualification threshold, the amount of cyclised impurity would breach the qualification threshold at 1 month for the 2 mg capsules. See Figure lc.
  • the lactose tablet formulation showed no increase in the amount of cyclised impurity within the first 2 months, demonstrating high stability even at the accelerated conditions, and is thus projected not to breach the qualification threshold of 0.5% within 6 months.
  • the qualification threshold For the 0.2% qualification threshold, no increase in the amount of cyclised impurit was seen within the first 4 months, and is thus projected not to breach the qualification threshold within 6 months. See Figures Id and le. Long-term Conditions
  • the lactose capsule formulation showed a very small increase in the amount of cyclised impurity in the 2 mg strength in the first month, whereas the tablet formulation again demonstrated no increase in the amount of cyclised impurity. See Figures 2c and 2d.
  • ORFADIN 5 mg and 10 mg capsules may achieve 12 months within specification at the long-term storage conditions, the accelerated conditions have demonstrated that they are highly sensitive to any increase in temperature due to increased cyclisation of the drug substance.
  • the tablet formulation of the present invention has demonstrated stability at both the long-term and accelerated conditions, with no formation of the cyclised impurity observed. This shows that a room temperature storage condition is appropriate and the pharmaceutical product is not sensitive to an increase in the temperature due to the storage conditions.
  • HPLC conditions were as follows:

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
PCT/GB2015/050006 2014-01-03 2015-01-05 Pharmaceutical composition WO2015101794A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
PL15700003T PL3089740T3 (pl) 2014-01-03 2015-01-05 Kompozycja farmaceutyczna
DK15700003.5T DK3089740T3 (da) 2014-01-03 2015-01-05 Farmaceutisk sammensætning
EP15700003.5A EP3089740B1 (en) 2014-01-03 2015-01-05 Pharmaceutical composition
US15/109,220 US10328029B2 (en) 2014-01-03 2015-01-05 Pharmaceutical composition
CA2935013A CA2935013C (en) 2014-01-03 2015-01-05 Pharmaceutical composition containing nitisinone, and its use
LTEP15700003.5T LT3089740T (lt) 2014-01-03 2015-01-05 Farmacinė kompozicija
ES15700003T ES2806739T3 (es) 2014-01-03 2015-01-05 Composición farmacéutica
RS20200849A RS60551B1 (sr) 2014-01-03 2015-01-05 Farmaceutska kompozicija
SI201531285T SI3089740T1 (sl) 2014-01-03 2015-01-05 Farmacevtski sestavek
CY20201100659T CY1123394T1 (el) 2014-01-03 2020-07-20 Φαρμακευτικη συνθεση
HRP20201135TT HRP20201135T1 (hr) 2014-01-03 2020-07-20 Farmaceutski pripravak

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1400117.6 2014-01-03
GBGB1400117.6A GB201400117D0 (en) 2014-01-03 2014-01-03 Pharmaceutical composition

Publications (1)

Publication Number Publication Date
WO2015101794A1 true WO2015101794A1 (en) 2015-07-09

Family

ID=50191740

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2015/050006 WO2015101794A1 (en) 2014-01-03 2015-01-05 Pharmaceutical composition

Country Status (15)

Country Link
US (1) US10328029B2 (pt)
EP (1) EP3089740B1 (pt)
CA (1) CA2935013C (pt)
CY (1) CY1123394T1 (pt)
DK (1) DK3089740T3 (pt)
ES (1) ES2806739T3 (pt)
GB (1) GB201400117D0 (pt)
HR (1) HRP20201135T1 (pt)
HU (1) HUE050881T2 (pt)
LT (1) LT3089740T (pt)
PL (1) PL3089740T3 (pt)
PT (1) PT3089740T (pt)
RS (1) RS60551B1 (pt)
SI (1) SI3089740T1 (pt)
WO (1) WO2015101794A1 (pt)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ITUB20160650A1 (it) * 2016-02-11 2017-08-11 Dipharma S A Formulazioni farmaceutiche solide stabili contenenti 2-(2-nitro-4-trifluorometilbenzoil)-1,3-cicloesandione
US10093610B2 (en) 2016-11-30 2018-10-09 Dipharma S.A. Crystalline inhibitor of 4-hydroxyphenylpyruvate dioxygenase

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993000080A1 (en) * 1991-06-24 1993-01-07 Zeneca Ltd. Use of 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione in the treatment of tyrosinaemia and pharmaceutical compositions
CN102976948A (zh) * 2012-11-28 2013-03-20 郑州大明药物科技有限公司 尼替西农的制备方法
WO2013181292A1 (en) * 2012-05-29 2013-12-05 Biotie Therapies, Inc. Nitisinone formulations

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IS2385B (is) * 2006-02-10 2008-07-15 Actavis Group Hf. Klópidógrel bísúlfat lyfjasamsetningar

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993000080A1 (en) * 1991-06-24 1993-01-07 Zeneca Ltd. Use of 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione in the treatment of tyrosinaemia and pharmaceutical compositions
WO2013181292A1 (en) * 2012-05-29 2013-12-05 Biotie Therapies, Inc. Nitisinone formulations
CN102976948A (zh) * 2012-11-28 2013-03-20 郑州大明药物科技有限公司 尼替西农的制备方法

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ITUB20160650A1 (it) * 2016-02-11 2017-08-11 Dipharma S A Formulazioni farmaceutiche solide stabili contenenti 2-(2-nitro-4-trifluorometilbenzoil)-1,3-cicloesandione
WO2017137468A1 (en) 2016-02-11 2017-08-17 Dipharma S.A. Stable solid pharmaceutical formulations containing 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione
CN108495623A (zh) * 2016-02-11 2018-09-04 迪法玛公司 含有2-(2-硝基-4-三氟甲基-苯甲酰基)-1,3-环己二酮的稳定固体药物配制品
US10888527B2 (en) 2016-02-11 2021-01-12 Dipharma S.A. Stable solid pharmaceutical formulations containing 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cylcohexanedione
CN108495623B (zh) * 2016-02-11 2021-04-20 迪法玛公司 含有2-(2-硝基-4-三氟甲基-苯甲酰基)-1,3-环己二酮的稳定固体药物配制品
US10093610B2 (en) 2016-11-30 2018-10-09 Dipharma S.A. Crystalline inhibitor of 4-hydroxyphenylpyruvate dioxygenase

Also Published As

Publication number Publication date
SI3089740T1 (sl) 2020-09-30
HRP20201135T1 (hr) 2021-01-22
PT3089740T (pt) 2020-07-24
US20160324785A1 (en) 2016-11-10
ES2806739T3 (es) 2021-02-18
RS60551B1 (sr) 2020-08-31
US10328029B2 (en) 2019-06-25
GB201400117D0 (en) 2014-02-19
EP3089740B1 (en) 2020-05-13
LT3089740T (lt) 2020-08-10
EP3089740A1 (en) 2016-11-09
PL3089740T3 (pl) 2020-11-02
CA2935013A1 (en) 2015-07-09
CA2935013C (en) 2022-08-02
HUE050881T2 (hu) 2021-01-28
CY1123394T1 (el) 2021-12-31
DK3089740T3 (da) 2020-08-03

Similar Documents

Publication Publication Date Title
JP5775464B2 (ja) 非晶質cddo−meを含有する遅延放出性経口投薬組成物
EP3501510A1 (en) Controlled release enteric soft capsules of fumarate esters
US20240075039A1 (en) Oral solid tablet comprising bruton's tyrosine kinase inhibitor and preparation method therefor
US9925208B2 (en) Compositions comprising S-adenosylmethionine and a gallic acid ester
US20190388434A1 (en) (17-ß)-3-Oxoandrost-4-En-17-Yl Undecanoate Compositions and Methods of Their Preparation and Use
US20230190732A1 (en) Pharmaceutical composition containing nitroxoline prodrug, and preparation method and application therefor
JP7241807B2 (ja) 15β-ヒドロキシ-酢酸オサテロンの結晶多形
JP2018519279A (ja) イリノテカン含有経口用固形製剤及びその製造方法
AU2018326596B2 (en) High concentration dosage forms of pridopidine
KR20090067210A (ko) 페닐알킬 카바메이트 조성물
US20190248830A1 (en) Proliposomal testosterone undecanoate formulations
TW201338777A (zh) 用於治療金屬過載之經口調配物
WO2017054741A1 (zh) 提高索拉非尼生物利用度的制剂
AU2015389109B2 (en) Enteric-coated pellets containing a proton pump inhibitor
CA2935013C (en) Pharmaceutical composition containing nitisinone, and its use
JP5678088B2 (ja) 4−メチルピラゾール製剤
CN103214382A (zh) 一种盐酸甲氯芬酯化合物及其药物组合物
RU2702746C1 (ru) Стабильные твёрдые фармацевтические составы, содержащие 2-(2-нитро-4-трифторметилбензоил)-1,3-циклогександион
US11013707B2 (en) Administration of oral methyldopa
KR101609105B1 (ko) 용출성 및/또는 흡수성이 개선된 경구 투여용 의약 조성물
BR112021001525A2 (pt) sal de malonato de epinefrina, composição farmacêutica, método para fabricar um sal de malonato de epinefrina e método para tratamento de um paciente que sofre de uma afecção alérgica
KR20220015437A (ko) 피리미디닐아미노-피라졸 화합물의 변형 방출 제제, 및 치료 방법
JP2020117478A (ja) イソクエルシトリン組成物
CZ21562U1 (cs) Kompozice tibolonu s definovanou velikosti částic

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15700003

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2935013

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 15109220

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015700003

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015700003

Country of ref document: EP